Jeon, You, and Park et al. detected the highest 4-1BB expression on terminally exhausted tumor-specific CD8+ TIL in human melanoma and hepatocellular carcinoma. These cells did not respond to pembrolizumab treatment, but ABL503, an anti-4-1BB×PD-L1 bispecific antibody, increased their in vitro proliferation, which was further enhanced in combination with anti-PD-1. While either monotherapy induced some tumor growth delay in a 4-1BB-, PD-1-, and PD-L1-humanized murine tumor model, combination treatment of anti-PD-1 and ABL503 further delayed tumor growth, with increased TIL density and activation of CD8+ TIL.

Contributed by Maartje Wouters

PURPOSE: To overcome the limited efficacy of immune checkpoint blockade, there is a need to find novel cancer immunotherapeutic strategies for the optimal treatment of cancer. The novel anti-4-1BB_PD-L1 bispecific antibody-ABL503 (also known as TJ-L14B)-was designed to simultaneously target PD-L1 and 4-1BB, and demonstrated strong antitumor T-cell responses without considerable toxicity. Here, we investigated how the combination of ABL503 and anti-PD-1 blockade affected the reinvigoration of exhausted tumor-infiltrating CD8+ T cells (CD8+ TILs) and anti-tumor efficacy. EXPERIMENTAL DESIGN: Single cell suspensions of hepatocellular carcinoma and ovarian cancer from treatment-naive patients were used for immunophenotyping of CD8+ TILs and in vitro functional assays. Humanized hPD-1/hPD-L1/h4-1BB triple knock-in mice were used to evaluate the effects of ABL503 and anti-PD-1 blockade in vivo. RESULTS: We observed that ABL503 successfully restored the functions of 4-1BB+ exhausted CD8+ TILs, which were enriched for tumor-specific T cells but unresponsive to anti-PD-1 blockade. Importantly, compared to anti-PD-1 blockade alone, the combination of ABL503 and anti-PD-1 blockade further enhanced the functional restoration of human CD8+ TILs in vitro. Consistently, the combination of ABL503 with anti-PD-1 in vivo significantly alleviated tumor growth, and induced enhanced infiltration and activation of CD8+ TILs. CONCLUSIONS: ABL503-a PD-L1 and 4-1BB dual-targeting bispecific antibody-elicits pronounced additive tumor growth inhibition, with increased infiltration and functionality of exhausted CD8+ T cells, which in turn enhances the anti-cancer effects of anti-PD-1 blockade. These promising findings suggest that ABL503 (TJ-L14B) in combination with PD-1 inhibitors will likely further enhance therapeutic benefit in clinical trials.

Author Info: (1) Seoul National University Bundang Hospital, Seongnam, Korea (South), Republic of. (2) ABL Bio Inc., Seongnam, Korea (South), Republic of. (3) Soonchunhyang University Bucheon H

Author Info: (1) Seoul National University Bundang Hospital, Seongnam, Korea (South), Republic of. (2) ABL Bio Inc., Seongnam, Korea (South), Republic of. (3) Soonchunhyang University Bucheon Hospital, Bucheon, Korea (South), Republic of. (4) Korea Advanced Institute of Science and Technology, Korea (South), Republic of. (5) ABL Bio Inc., Seongnam, Korea (South), Republic of. (6) ABL Bio, Korea (South), Republic of. (7) ABL Bio, Korea (South), Republic of. (8) ABL Bio, Korea (South), Republic of. (9) Asan Institute for Life Sciences, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Korea (South), Republic of. (10) Asan Medical Center, Seoul, Korea (South), Republic of. (11) Korea Advanced Institute of Science and Technology, Daejeon, Korea (South), Republic of. (12) Yonsei University College of Medicine, Seoul, Korea (South), Republic of. (13) Yonsei University college of medicine, Seoul, Korea (South), Republic of. (14) ABL Bio, Seongnam, Korea (South), Republic of. (15) Korea Advanced Institute of Science and Technology, Daejeon, Korea (South), Republic of.